Language selection

Search

Patent 2872188 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2872188
(54) English Title: METHODS AND COMPOSITIONS FOR TREATMENT OF TRAUMATIC BRAIN INJURY AND FOR MODULATION OF MIGRATION OF NEUROGENIC CELLS
(54) French Title: PROCEDES ET COMPOSITIONS DESTINES AU TRAITEMENT D'UNE LESION CEREBRALE TRAUMATIQUE ET A LA MODULATION DE LA MIGRATION DE CELLULES NEUROGENES
Status: Granted and Issued
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 35/28 (2015.01)
  • A61P 25/00 (2006.01)
  • C12N 5/0775 (2010.01)
  • C12N 5/079 (2010.01)
  • C12N 5/10 (2006.01)
(72) Inventors :
  • BORLONGAN, CESAR V. (United States of America)
  • CASE, CASEY C. (United States of America)
(73) Owners :
  • UNIVERSITY OF SOUTH FLORIDA
  • SANBIO, INC.
(71) Applicants :
  • UNIVERSITY OF SOUTH FLORIDA (United States of America)
  • SANBIO, INC. (United States of America)
(74) Agent: KIRBY EADES GALE BAKER
(74) Associate agent:
(45) Issued: 2018-08-14
(86) PCT Filing Date: 2013-03-13
(87) Open to Public Inspection: 2013-11-21
Examination requested: 2014-11-14
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2013/030895
(87) International Publication Number: US2013030895
(85) National Entry: 2014-10-30

(30) Application Priority Data:
Application No. Country/Territory Date
61/647,893 (United States of America) 2012-05-16

Abstracts

English Abstract

Disclosed herein are methods for the treatment of traumatic brain injury by transplantation of cells descended from marrow adherent stem cells that express an exogenous Notch intracellular domain. The transplanted cells form a pathway along which endogenous neurogenic cells proliferate and migrate from the subventricular zone to the site of injury.


French Abstract

La présente invention concerne des procédés de traitement d'une lésion cérébrale traumatique par transplantation de cellules descendues de cellules souches adhérentes de moelle qui expriment un domaine intracellulaire de Notch exogène. Les cellules transplantées forment une voie le long de laquelle des cellules neurogènes endogènes prolifèrent et migrent de la zone sous-ventriculaire vers le site de lésion.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. Cells for use to stimulate proliferation of endogenous neurogenic cells in
a
subject in order to treat a traumatic brain injury in the subject, wherein
said cells are
obtained by a process comprising the steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences encoding a Notch intracellular domain (NICD), wherein said
polynucleotide does not encode a full-length Notch protein,
(c) selecting cells that comprise the polynucleotide of step (b), and
(d) further culturing the selected cells of step (c) in the absence of
selection.
2. The cells of claim 1, wherein the subject is a human.
3. The cells of claim 1 or claim 2, wherein the MSCs are obtained from a
human.
4. Cells for use to induce migration of endogenous neurogenic cells from a
neurogenic niche to a site of a traumatic brain injury in a subject, in order
to treat the
traumatic brain injury, wherein said cells are obtained by a process
comprising the
steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences encoding a Notch intracellular domain (NICD), wherein said
polynucleotide does not encode a full-length Notch protein,
(c) selecting cells that comprise the polynucleotide of step (b). and
(d) further culturing the selected cells of step (c) in the absence of
selection.
5. The cells of claim 4, wherein the neurogenic niche is a subventricular
zone.
6. The cells of claim 4 or 5, wherein the subject is a human.
7. The cells of claims 4, 5 or 6, wherein the MSCs are obtained from a
human.
29

8. Use of cells to stimulate proliferation of endogenous neurogenic cells in a
subject, for the treatment of a traumatic brain injury in the subject, wherein
said cells
are obtained by a process comprising the steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences encoding a Notch intracellular domain (NICD), wherein said
polynucleotide does not encode a full-length Notch protein,
(c) selecting cells that comprise the polynucleotide of step (b), and
(d) further culturing the selected cells of step (c) in the absence of
selection.
9. The use of claim 8, wherein the subject is a human.
10. The use of claim 8 or claim 9, wherein the MSCs are obtained from a
human.
11. Use of cells in a subject with a traumatic brain injury, to induce
migration
of endogenous neurogenic cells from a neurogenic niche to a site of the
traumatic
brain injury, for the treatment of the brain injury, wherein said cells are
obtained by a
process comprising the steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences encoding a Notch intracellular domain (NICD), wherein said
polynucleotide does not encode a full-length Notch protein,
(c) selecting cells that comprise the polynucleotide of step (b), and
(d) further culturing the selected cells of step (c) in the absence of
selection.
12. The use of claim 11, wherein the neurogenic niche is a subventricular
zone.
13. The use of claim 11 or 12, wherein the subject is a human.
14. The use of claim 11, 12 or 13, wherein the MSCs are obtained from a
human.

15. Use of cells in the manufacture of a medicament for treating a traumatic
brain injury in a subject by stimulating proliferation of endogenous
neurogenic cells
in the subject, wherein said cells are obtained by a process comprising the
steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences encoding a Notch intracellular domain (N1CD) wherein said
polynucleotide
does not encode a full-length Notch protein,
(c) selecting cells that comprise the polynucleotide of step (b), and
(d) further culturing the selected cells of step (c) in the absence of
selection.
16. The use of claim 15, wherein the subject is a human.
17. The use of claim 15 or claim 16, wherein the MSCs are obtained from a
human.
18. Use of cells in the manufacture of a medicament for treating a traumatic
brain injury in a subject by inducing migration of endogenous neurogenic cells
from a
neurogenic niche to a site of the traumatic brain injury, wherein said cells
are obtained
by a process comprising the steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences encoding a Notch intracellular domain (NICD) wherein said
polynucleotide
does not encode a full-length Notch protein,
(c) selecting cells that comprise the polynucleotide of step (b), and
(d) further culturing the selected cells of step (c) in the absence of
selection.
19. The use of claim 18, wherein the neurogenic niche is a subventricular
zone.
20. The use of claim 18 or 19, wherein the subject is a human.
21. The use of claim 18, 19 or 20, wherein the MSCs are obtained from a
human.
31

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02872188 2016-05-09
METHODS AND COMPOSITIONS FOR TREATMENT OF
TRAUMATIC BRAIN INJURY AND FOR
MODULATION OF MIGRATION OF NEUROGENIC CELLS
[0001]
[0002]
FIELD
[0003] The present disclosure is in the field of cell therapy for
neurological
disorders.
BACKGROUND
[0004] Initially employed for in-depth examination of cell development',
stem
cells have become a cornerstone for regenerative medicine, including cell-
based
therapies for treatment of neurological disorders2'3. Stem cells exist even in
adulthood8, and possess the capacity to self-renew and differentiate into
multiple
lineages9, contribute to normal homeostasie, and exert therapeutic benefits
either
endogenously11-14 or following transplantation in injured organs, i.e.,
bra1n15-21. The
subventricular zone (SVZ) of the lateral ventricles and the subgranular zone
of the
hippocampus dentate gyms are the two major stem-cell niches in the adult
brain22'23,
although quiescent neural stem cells (NSCs) have been detected in other brain
regions24. Induction of endogenous stem cells after injury would provide new
1-
opportunities in regenerative medicine2 '3 121 .
[0005] Cells other than pluripotent stem cells have also been used in
the
treatment of disorders of the central nervous system. As one example, SB623
cells
(which are cells derived from marrow adherent stem cells in which an exogenous
Notch intracellular domain has been expressed) are used for the treatment of
stroke,
1

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
by transplantation at or near the site of ischemic insult. See, for example,
U.S. Patent
No. 8,092,792 and Yasuhara et a/. (2009) Stem Cells Devel. 18:1501-1513. U.S.
Patent No. 7,682,825 describes additional uses of SB623 cells in the treatment
of a
number of disorders of the central and peripheral nervous systems.
[0006] Despite these scientific advances and some initial clinical
studies25-27 , a
fundamental gap in our understanding of cell therapy is a knowledge of the
mechanisms by which transplanted cells facilitate the repair of damaged neural
tissue.
To date, increased graft survival and graft persistence have been considered
the crux
of successful cell transplantation therapy in affording therapeutic benefits
in
hematologic and non-hematologic disorders. Thus, much effort has been directed
to
prolonging the survival and persistence of transplanted cells. Accordingly,
methods
for effective cell therapy, that do not require the persistence of large
amounts of
transplanted cells, would be advantageous.
[0007] Traumatic brain injury (FBI) refers to damage to the brain
resulting
from external mechanical force. TBI can result from falls, firearm wounds,
sports
accidents, construction accidents and vehicle accidents, among other causes.
Victims
of TBI can suffer from a number of physical, cognitive, social, emotional
and/or
behavioral disorders.
[0008] Little can be done to reverse the initial physical damage of a
TBI.
Therefore, treatment options consist primarily of stabilization to prevent
further
damage in the acute phase, and rehabilitation thereafter. Because of these
limited
options, additional methods and compositions for treatment of TBI are needed.
SUMMARY
[0009] The present inventors have discovered that transplantation of SB623
cells (i.e., cells derived from marrow adherent stem cells in which an
exogenous
Notch intracellular domain has been expressed) can be used in the treatment of
traumatic brain injury (TBI). Animals that received transplants of SB623 cells
after
TBI displayed significantly improved motor and neurological functions coupled
with
significantly reduced damage to the cortical core and pen-injured cortical
areas,
compared to traumatically injured animals that received injection of vehicle
only.
[0010] The inventors have also found that, contrary to expectations,
survival
and persistence of large numbers of transplanted cells are not required for
the
therapeutic benefits of SB623 cell transplantation. Surprisingly, therapeutic
benefits
2

I I
CA 2872188 2017-04-25
can be obtained by minimum and acute graft survival, which is sufficient to
initiate a
robust and stable functional recovery. This solves two major problems: the
need for
an ample supply of transplantable cells and the need for long-term graft
survival.
[0011] The inventors have also discovered that the beneficial effects
of SB623
cell transplantation, in the treatment of TBI, result from the formation of a
biological
bridge ("biobridge") between the neurogenic niche in the subventricular zone
(SVZ)
and the injured brain site. This biobridge, which has been visualized
immunohistochemically and laser-captured, initially expressed high levels of
extracellular matrix metalloproteinases and was characterized by a stream of
the
transplanted cells. At later times after transplantation, the grafted cells
were replaced
by newly formed host cells, and few-to-no transplanted cells remained in the
biobridge. Thus, the transplanted SB623 cells initially formed a pathway
between the
neurogenic SVZ and the injured cortex that facilitated later migration of host
neurogenic cells from the neurogenic niche to the site of brain injury.
[0012] This sequence of events reveals a novel method for treatment of TBI;
namely, transplantation of SB623 cells, which form transient pathways for
directing
the migration of host neurogenic cells. That is, the transplanted SB623 cells
initially
form a biobridge between a neurogenic niche and the site of injury; but once
this
biobridge is formed, the grafted cells are replaced by host neurogenic cells
which
migrate to the injury site. These findings reveal that long-distance migration
of host
cells from a neurogenic niche to an injured brain site can be achieved through
transplanted SB623 cells serving as biobridges for initiation of endogenous
repair
mechanisms.
[0012a] Certain exemplary embodiments provide cells for use to
stimulate
proliferation of endogenous neurogenic cells in a subject in order to treat a
traumatic
brain injury in the subject, wherein said cells are obtained by a process
comprising the
steps of: (a) providing a cell culture of marrow adherent stem cells (MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences
encoding a Notch intracellular domain (NICD), wherein said polynucleotide does
not
encode a full-length Notch protein, (c) selecting cells that comprise the
polynucleotide
of step (b), and (d) further culturing the selected cells of step (c) in the
absence of
selection.
[0012b] Other exemplary embodiments provide cells for use to induce
migration of endogenous neurogenic cells from a neurogenic niche to a site of
a
3

CA 2872188 2017-04-25
traumatic brain injury in a subject, in order to treat the traumatic brain
injury, wherein
said cells are obtained by a process comprising the steps of: (a) providing a
cell
culture of marrow adherent stem cells (MSCs), (b) contacting the cell culture
of
step (a) with a polynucleotide comprising sequences encoding a Notch
intracellular
domain (NICD), wherein said polynucleotide does not encode a full-length Notch
protein, (c) selecting cells that comprise the polynucleotide of step (b), and
(d) further
culturing the selected cells of step (c) in the absence of selection.
100120 Yet other exemplary embodiments provide use of cells to
stimulate
proliferation of endogenous neurogenic cells in a subject, for the treatment
of a
traumatic brain injury in the subject, wherein said cells are obtained by a
process
comprising the steps of: (a) providing a cell culture of marrow adherent stem
cells
(MSCs), (b) contacting the cell culture of step (a) with a polynucleotide
comprising
sequences encoding a Notch intracellular domain (NICD), wherein said
polynucleotide does not encode a full-length Notch protein, (c) selecting
cells that
comprise the polynucleotide of step (b), and (d) further culturing the
selected cells of
step (c) in the absence of selection.
[0012d] Still yet other exemplary embodiments provide use of cells in a
subject
with a traumatic brain injury, to induce migration of endogenous neurogenic
cells
from a neurogenic niche to a site of the traumatic brain injury, for the
treatment of the
brain injury, wherein said cells are obtained by a process comprising the
steps of:
(a) providing a cell culture of marrow adherent stem cells (MSCs), (b)
contacting the
cell culture of step (a) with a polynucleotide comprising sequences encoding a
Notch
intracellular domain (NICD), wherein said polynucleotide does not encode a
full-
length Notch protein, (c) selecting cells that comprise the polynucleotide of
step (b),
and (d) further culturing the selected cells of step (c) in the absence of
selection.
[0012e] Still yet other exemplary embodiments provide use of cells in
the
manufacture of a medicament for treating a traumatic brain injury in a subject
by
stimulating proliferation of endogenous neurogenic cells in the subject,
wherein said
cells are obtained by a process comprising the steps of: (a) providing a cell
culture of
marrow adherent stem cells (MSCs), (b) contacting the cell culture of step (a)
with a
polynucleotide comprising sequences encoding a Notch intracellular domain
(NICD)
wherein said polynucleotide does not encode a full-length Notch protein, (c)
selecting
cells that comprise the polynucleotide of step (b), and (d) further culturing
the
selected cells of step (c) in the absence of selection.
3a

I I
CA 2872188 2017-04-25
1001211 Still yet other exemplary embodiments provide use of cells in
the
manufacture of a medicament for treating a traumatic brain injury in a subject
by
inducing migration of endogenous neurogenic cells from a neurogenic niche to a
site
of the traumatic brain injury, wherein said cells are obtained by a process
comprising
the steps of: (a) providing a cell culture of marrow adherent stem cells
(MSCs),
(b) contacting the cell culture of step (a) with a polynucleotide comprising
sequences
encoding a Notch intracellular domain (NICD) wherein said polynucleotide does
not
encode a full-length Notch protein, (c) selecting cells that comprise the
polynucleotide
of step (b), and (d) further culturing the selected cells of step (c) in the
absence of
selection.
[0013] Accordingly, the present disclosure provides, inter alia, the
following
embodiments:
1. A method for treating traumatic brain injury in a subject, the method
comprising administering, to the brain of the subject, a therapeutically
effective
amount of SB623 cells, wherein the SB623 cells are obtained by (a) providing a
culture of marrow adherent stem cells (MSCs), (b) contacting the cell culture
of step
(a) with a polynucleotide comprising sequences encoding a Notch intracellular
domain (NICD) wherein said polynucleotide does not encode a full-length Notch
protein, (c) selecting cells that comprise the polynucleotide of step (b), and
(d) further
culturing the selected cells of step (c) in the absence of selection.
2. The method of embodiment 1, wherein the subject is a human.
3b

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
3. The method of either of embodiments 1 or 2, wherein the MSCs are
obtained from a human.
4. Cells for transplantation into a subject for the treatment of traumatic
brain
injury, wherein said cells are obtained by a process comprising the steps of:
(a)
providing a culture of MSCs, (b) contacting the cell culture of step (a) with
a
polynucleotide comprising sequences encoding a NICD wherein said
polynucleotide
does not encode a full-length Notch protein, (c) selecting cells that comprise
the
polynucleotide of step (b), and (d) further culturing the selected cells of
step (c) in the
absence of selection.
5. The cells of embodiment 4, wherein the subject is a human.
6. The cells of either of embodiments 4 or 5, wherein the MSCs are obtained
from a human.
7. A method for inducing the migration of endogenous ncurogcnic cells from
a neurogenic niche to a site of brain injury, the method comprising
administering, to
the brain of a subject, a therapeutically effective amount of SB623 cells,
wherein the
SB623 cells are obtained by (a) providing a culture of MSCs, (b) contacting
the cell
culture of step (a) with a polynucleotide comprising sequences encoding a
Notch
intracellular domain (NICD) wherein said polynucleotide does not encode a full-
length Notch protein, (c) selecting cells that comprise the polynucleotide of
step (b),
and (d) further culturing the selected cells of step (c) in the absence of
selection.
8. The method of embodiment 7, wherein the neurogenic niche is the
subventricular zone.
9. The method of either of embodiments 7 or 8, wherein the brain injury is a
traumatic brain injury.
10. The method of any of embodiments 7-9, wherein the subject is a human.
11. The method of any of embodiments 7-10, wherein the MSCs are obtained
from a human.
12. A method for stimulating proliferation of neurogenic cells in a subject,
the
method comprising administering, to the brain of a subject, a therapeutically
effective
amount of SB623 cells, wherein the SB623 cells are obtained by (a) providing a
culture of MSCs, (b) contacting the cell culture of step (a) with a
polynucleotide
comprising sequences encoding a Notch intracellular domain (NICD) wherein said
polynucleotide does not encode a full-length Notch protein, (c) selecting
cells that
4

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
comprise the polynucleotide of step (b), and (d) further culturing the
selected cells of
step (c) in the absence of selection.
13. The method of embodiment 12, wherein the brain injury is a traumatic
brain injury.
14. The method of either of embodiments 12 or 13, wherein the subject is a
human.
15. The method of any of embodiments 12-14, wherein the MSCs are
obtained from a human.
16. A method for inducing neurogenic cells to proliferate and migrate to a
site
of brain injury in a subject, the method comprising administering, to the
brain of a
subject, a therapeutically effective amount of SB623 cells, wherein the SB623
cells
are obtained by (a) providing a culture of MSCs, (b) contacting the cell
culture of step
(a) with a polynucleotide comprising sequences encoding a Notch intracellular
domain (NICD) wherein said polynucleotide does not encode a full-length Notch
protein, (c) selecting cells that comprise the polynucleotide of step (b), and
(d) further
culturing the selected cells of step (c) in the absence of selection.
17. The method of embodiment 16, wherein the brain injury is a traumatic
brain injury.
18. The method of either of embodiments 16 or 17, wherein the subject is a
human.
19. The method of any of embodiments 16-18, wherein the MSCs are
obtained from a human.
BRIEF DESCRIPTION OF THE DRAWINGS
[0014] Figure 1 shows results of the elevated body swing test (EBST)
in rats.
Values are provided for Baseline (before TBI) and for 7 days, 1 month, 2
months and
3 months after TBI. The rats received either transplants of SB623 cells
("Cells,"
right-most bar in each pair) or infusion of vehicle ("Vehicle," left-most bar
in each
pair). "*" indicates statistical significance with a p < 0.05.
[0015] Figure 2 shows mean scores in a modified Bederson neurological
examination in rats subjected to FBI that subsequently received either
transplants of
SB623 cells ("Cells") or infusion of vehicle ("Vehicle"). Values are provided
for
Baseline (before TBI) and for 7 days, 1 month, 2 months and 3 months after
TBI. The
5

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
left-most bar in each pair represents the score in rats infused with vehicle;
the right-
most bar in each pair represents the score in rats that received transplants
of SB623
cells. "*" indicates statistical significance with a p <0.05.
[0016] Figure 3 shows mean values for the number of seconds rats were
able
to remain on a Rotorod apparatus. The rats were subjected to experimental TBI
and
subsequently received either transplants of SB623 cells ("Cells") or infusion
of
vehicle ("Vehicle"). Values are provided for Baseline (before TBI) and for 7
days, 1
month, 2 months and 3 months after TBI. The left-most bar in each pair
represents
the score in rats infused with vehicle; the right-most bar in each pair
represents the
score in rats that received transplants of SB623 cells. "*" indicates
statistical
significance with a p <0.05.
[0017] Figures 4A and 4B shows results of assays for damage to the
cortical
core ("Core") and to the cortical region in and around the impact site ("Pen-
injury")
in rats subjected to TBI. Figure 4A shows H&E sections of brains from rats
that
received transplants of SB623 cells (panels a-d) compared to rats that
received
infusion of vehicle (panels al-di). In Figure 4B, the results are expressed as
percent
lesioned area (see Example 8) relative to animals subjected to TBI that
received
infusion of vehicle. The left-most bar in each pair shows values for the core
region;
the right-most bar in each pair shows values for the pen-injury region.
[0018] Figure 5 shows levels of Ki67-labeled cells in the subventricular
zone
("SVZ") and the cortex ("CTX") of animals subjected to TBI that received
transplants
of SB623 cells, compared to animals subjected to TBI that received infusions
of
vehicle, at one month and three months after TBI. "*" indicates a
statistically
significant increase in the number of labeled cell observed per high-power
field (p <
0.05).
[0019] Figure 6 shows levels of nestin-labeled cells in the
subventricular zone
("SVZ") and the cortex ("CTX") of animals subjected to TBI that received
transplants
of SB623 cells, compared to animals subjected to TBI that received infusions
of
vehicle, at one month and three months after TBI. "*" indicates a
statistically
significant increase in the number of labeled cell observed per high-power
field (p <
0.05).
[0020] Figure 7 shows levels of doubleeortin-labeled cells in the
corpus
callo sum ("CC") and the cortex ("CTX") of animals subjected to TBI that
received
transplants of SB623 cells, compared to animals subjected to TBI that received
6

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
infusions of vehicle, at one month and three months after TBI. "*" indicates a
statistically significant increase in the number of labeled cell observed per
high-power
field (p < 0.05).
[00211 Figure 8 shows lytic activity in homogenates of laser-captured
biobridges from the brains of rats subjected to experimental TBI, at one month
and
three months after TBI. Activities are expressed as optical density units,
relative to
0.5 ng recombinant MMP-9, obtained by scanning zymographic gels. In each of
the
two sets of three bars, the left-most bar represents relative activity in
biobridges from
rats infused with vehicle after TBI, the center bar represents relative
activity in
biobridges from rats transplanted with SB623 cells after TBI, and the right-
most bar
represents relative activity in biobridges from control, sham-operated rats.
DETAILED DESCRIPTION
[0022] Disclosed herein are methods and compositions for treatment of
traumatic brain injury (TBI). Also disclosed herein are methods and
compositions for
modulation of the migration of stem cells (e.g., neural stem cells, neuronal
stem cells)
in the brain.
[0023] The inventors have made the surprising discovery that the
behavioral
and histological improvements resulting from cell transplantation, after TBI,
do not
require large-scale graft survival or long-term graft persistence. Indeed,
only modest,
acute graft survival is necessary to produce these therapeutic benefits. Thus,
the
inventors have uncovered a novel method for neural repair that entails a
threshold
dose of transplanted cells, which do not need to persist in the brain, and
which are
capable of inducing the SVZ to generate and propel new cells to the impacted
cortical
area. Accordingly, transplanting the minimum effective dose and the acute
survival
of the transplanted cells are sufficient to initiate an intricate endogenous
restorative
machinery for abrogating a massive brain injury.
[0024] Practice of the present disclosure employs, unless otherwise
indicated,
standard methods and conventional techniques in the fields of cell biology,
toxicology, molecular biology, biochemistry, cell culture, immunology,
neurology,
surgery, recombinant DNA and related fields as are within the skill of the
art. Such
techniques are described in the literature and thereby available to those of
skill in the
art. See, for example, Alberts, B. et al., "Molecular Biology of the Cell,"
5th edition.
Garland Science, New York, NY, 2008; Voet, D. et al. "Fundamentals of
7

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
Biochemistry: Life at the Molecular Level," 3"1 edition, John Wiley & Sons,
Hoboken, NJ, 2008; Sambrook, J. et al., "Molecular Cloning: A Laboratory
Manual,"
3rd edition, Cold Spring Harbor Laboratory Press, 2001; Ausubel, F. et al.,
"Current
Protocols in Molecular Biology," John Wiley & Sons, New York, 1987 and
periodic
updates; Freshney, RI., "Culture of Animal Cells: A Manual of Basic
Technique,"
4th edition, John Wiley & Sons, Somerset, NJ, 2000; and the series "Methods in
Enzymology," Academic Press, San Diego, CA.
Marrow adherent Stem Cells (MSCs)
[0025] The present disclosure provides methods for treating TBI and
modulating stem cell migration by transplanting SB623 cells to a site of brain
injury
in a subject. SB623 cells are obtained from marrow adherent stem cells (MSCs),
also
known as marrow adherent stromal cells and mesenchymal stem cells, by
expressing
the intracellular domain of the Notch protein in the MSCs. MSCs are obtained
by
selecting adherent cells (i.e., cells that adhere to tissue culture plastic)
from bone
marrow.
[0026] Exemplary disclosures of MSCs are provided in U.S. patent
application publication No. 2003/0003090; Prockop (1997) Science 276:71-74 and
Jiang (2002) Nature 418:41-49. Methods for the isolation and purification of
MSCs
can be found, for example, in U.S. Patent No. 5,486,359; Pittenger et al.
(1999)
Science 284:143-147 and Dezawa et al. (2001) Eur. J Neurosci. 14:1771-1776.
Human MSCs are commercially available (e.g., BioWhittaker, Walkersville, MD)
or
can be obtained from donors by, e.g., bone marrow aspiration, followed by
selection
for adherent bone marrow cells. See, e.g., WO 2005/100552.
[0027] MSCs can also be isolated from umbilical cord blood. See, for
example, Campagnoli et al. (2001) Blood 98:2396-2402; Erices et al. (2000) Br.
.1
Haematol. 109:235-242 and Hou et al. (2003) Int. J. Hematol. 78:256-261.
Additional sources of MSCs include, for example, menstrual blood and placenta.
Notch Intracellular Domain
[0028] The Notch protein is a transmembrane receptor, found in all
metazoans, that influences cell differentiation through intracellular
signaling. Contact
of the Notch extracellular domain with a Notch ligand (e.g, Delta, Serrate,
Jagged)
results in two proteolytic cleavages of the Notch protein, the second of which
is
8

CA 02872188 2016-05-09
catalyzed by a y-secretase and releases the Notch intracellular domain (NICD)
into the
cytoplasm. In the mouse Notch protein, this cleavage occurs between amino
acids
gly1743 and val1744. The NICD translocates to the nucleus, where it acts as a
transcription factor, recruiting additional transcriptional regulatory
proteins (e.g.,
MAM, histone acetylases) to relieve transcriptional repression of various
target genes
(e.g., Hes 1).
[0029] Additional details and information regarding Notch signaling
are
found, for example in Artavanis-Tsakonas et al. (1995) Science 268:225-232;
Mumm
and Kopan (2000) Develop. Biol. 228:151-165 and Ehebauer et al. (2006) Sci.
STKE
2006 (364), cm7. [DOI: 10.1126/stke.3642006cm7].
Cell Culture and Transfection
[0030] Standard methods for cell culture are known in the art. See,
for
example, R. I. Freshney "Culture of Animal Cells: A Manual of Basic
Technique,"
Fifth Edition, Wiley, New York, 2005.
[0031] Methods for introduction of exogenous DNA into cells (i.e.,
transfection), and methods for selection of cells comprising exogenous DNA,
are also
well-known in the art. See, for example, Sambrook et al. "Molecular Cloning: A
Laboratory Manual," Third Edition, Cold Spring Harbor Laboratory Press, 2001;
Ausubel et al., "Current Protocols in Molecular Biology," John Wiley & Sons,
New
York, 1987 and periodic updates.
SB623 Cells
[0032] In one embodiment for the preparation of SB623 cells, a culture
of
MSCs is contacted with a polynucleotide comprising sequences encoding a Notch
intracellular domain (NICD); e.g., by transfection; followed by enrichment of
transfected cells by drug selection and further culture. See, for example,
U.S. Patent
No. 7,682,825 (March 23, 2010); U.S. Patent Application Publication No.
2010/0266554 (Oct. 21, 2010); and WO 2009/023251 (Feb. 19, 2009); to which the
reader is directed to for the purposes of describing isolation of marrow
adherent stem
cells and conversion of marrow adherent stem cells to SB623 cells (denoted
"neural
precursor cells" and "neural regenerating cells" in those documents).
9

CA 02872188 2016-05-09
[0033] In these methods, any polynucleotide encoding a Notch
intracellular
domain (e.g., vector) can be used, and any method for the selection and
enrichment of
transfected cells can be used. For example, in certain embodiments, MSCs are
transfected with a vector containing sequences encoding a Notch intracellular
domain
and also containing sequences encoding a drug resistance marker (e.g.
resistance to
G418). In additional embodiments, two vectors, one containing sequences
encoding a
Notch intracellular domain and the other containing sequences encoding a drug
resistance marker, are used for transfection of MSCs. In these embodiments,
selection
is achieved, after transfection of a cell culture with the vector or vectors,
by adding a
selective agent (e.g., G418) to the cell culture in an amount sufficient to
kill cells that
do not comprise the vector but spare cells that do. Absence of selection
entails
removal of said selective agent or reduction of its concentration to a level
that does not
kill cells that do not comprise the vector. Following selection (e.g., for
seven days) the
selective agent is removed and the cells are further cultured (e.g., for two
passages).
[0034] Preparation of SB623 cells thus involves transient expression
of an
exogenous Notch intracellular domain in a MSC. To this end, MSCs can be
transfected with a vector comprising sequences encoding a Notch intracellular
domain
wherein said sequences do not encode a full-length Notch protein. All such
sequences
are well known and readily available to those of skill in the art. For
example, Del
Amo et al. (1993) Genomics 15:259-264 present the complete amino acid
sequences
of the mouse Notch protein; while Mumm and Kopan (2000) Devel. Biol. 228:151-
165 provide the amino acid sequence, from mouse Notch protein, surrounding the
so-
called S3 cleavage site which releases the intracellular domain. Taken
together, these
references provide the skilled artisan with each and every peptide containing
a Notch
intracellular domain that is not the full-length Notch protein; thereby also
providing
the skilled artisan with every polynucleotide comprising sequences encoding a
Notch
intracellular domain that does not encode a full-length Notch protein. The
foregoing
documents (Del Amo and Mumm) can be referred to for the purpose of disclosing
the
amino acid sequence of the full-length Notch protein and the amino acid
sequence of
the Notch intracellular domain, respectively.
[0035] Similar information is available for Notch proteins and nucleic
acids
from additional species, including rat, Xenopus, Drosophila and human. See,
for
example, Weinmaster et al. (1991) Development 113:199-205; Schroeter etal.
(1998)

CA 02872188 2016-05-09
Nature 393:382-386; NCBI Reference Sequence No. NM_017167 (and references
cited therein); SwissProt P46531 (and references cited therein); SwissProt
Q01705
(and references cited therein); and GenBank CAB40733 (and references cited
therein). The foregoing references can be referred to for the purpose of
disclosing the
amino acid sequence of the full-length Notch protein and the amino acid
sequence of
the Notch intracellular domain in a number of different species.
[0036] In additional embodiments, SB623 cells are prepared by
introducing,
into MSCs, a nucleic acid comprising sequences encoding a Notch intracellular
domain such that the MSCs do not express exogenous Notch extracellular domain.
Such can be accomplished, for example, by transfecting MSCs with a vector
comprising sequences encoding a Notch intracellular domain wherein said
sequences
do not encode a full-length Notch protein.
[0037] Additional details on the preparation of SB623 cells, and
methods for
making cells with properties similar to those of SB623 cells which can be used
in the
methods disclosed herein, are found in U.S. Patent No. 7,682,825; U.S. Patent
No.
8,133,725; and U.S. Patent Application Publication Nos. 2010/0266554 and
2011/0229442. Such references can be referred to for the purposes of providing
additional details on, and alternative methods for the preparation of, SB623
cells, and
for providing methods for making cells with properties similar to those of
SB623
cells. See also Dezawa et al. (2004) J. Clin. Invest. 113:1701-1710.
Reversal of Symptoms of TBI by Transplantation of SB623 Cells
[0038] The efficacy of SB623 cell transplantation as a treatment for
TBI was
tested in a rat model system. Prior to testing, adult male Sprague-Dawley rats
(8-
weeks old) were evaluated in motor and neurological tests (all performed by
two
investigators blinded to the treatment condition throughout the study) to
confirm that
all animals displayed normal behaviors at baseline (i.e., prior to brain
insult).
Animals were then exposed to experimental traumatic brain injury (TBI), and
seven
days later were subjected to the same behavioral tests to confirm the typical
TBI-
induced motor and neurological impairments. Following these tests (at 7 days
post-
TBI), the animals were assigned randomly to one of two groups to receive
either
11

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
stereotaxic transplants of Notch-induced bone marrow-derived stem cells (SB623
cells)26-29 or vehicle infusion into the cortex (see Example 3).
10039] The inventors have found that, at both one month and three
months
post-TBI, traumatically injured animals that received transplants of SB623
cells
displayed significantly improved motor and neurological functions, coupled
with
significantly reduced damage to the cortical core and pen-injured cortical
areas,
compared to traumatically injured animals that received vehicle only (see
Examples).
These behavioral and physical improvements were achieved with modest graft
survival of 0.60% and 0.16% at one month and three months post-TBI,
respectively.
Other sites in the brain that are affected by TBI include the striatum and the
hippocampus; hence transplantation of SB623 cells to the striatum and the
hippocampus can also be used for treatment of TB1 affecting these areas. In
summary, transplantation of SB623 cells into brain-injured animals provided
robust
functional recovery despite lack of graft persistence.
Creation of a Biobridge by Transplantation of SB623 Cells
[0040] Examination of host tissue in brain-injured animals that had
received
transplants of SB623 cells, at one month post-TBI, revealed a surge of
endogenous
cell proliferation (detected by Ki67 expression) and differentiation of
neurogenic cells
(detected by expression of nestin) in the pen-injured cortical areas and
subventricular
zone (SVZ). A stream of cells (expressing doublecortin) migrating along the
corpus
callosum (CC) of these animals was also detected. In contrast, animals
subjected to
experimental TBI that received vehicle alone displayed limited cell
proliferation, little
neural differentiation, and only scattered migration in the pen-injured
cortical areas.
In addition, very few newly-foimed cells were visible in the sub-ventricular
zone of
these control animals (see Examples).
[0041] At three months post-TBI, the brains from animals that had
received
SB623 cell transplants exhibited much higher levels of cell proliferation and
neural
differentiation encasing the pen-injured cortical areas, along with a solid
stream of
neuronal cells (expressing both nestin and doublecortin) migrating not just
along but
across the CC from the SVZ to the impacted cortex. Brains from injured animals
that
had received only vehicle exhibited much more elevated levels of cell
proliferation at
three months post-TBI than at one month post-TBI, but the newly-formed cells
appeared "trapped" within the SVZ and the corpus callosum; with only a small
12

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
number of cells able to reach the impacted cortex. Quantitative analysis of
Ki67,
nestin and doublecortin immunoreactivity in the SVZ, the CC, and the injured
cortical
area indicated that the differences in expression of these markers, between
animals
receiving SB623 cell transplants and animals receiving only vehicle, were
statistically
significant.
[0042] In a separate experiment, the biobridge foimed by endogenous
cells
migrating from the SVZ to the site of injury was isolated by laser capture
microdissection (Espina et al. (2006) Nature Protoc. 1:586-603) and its
zymogenic
properties were analyzed. In this experiment, three groups of animals were
analyzed:
(1) animals subjected to TBI followed by transplantation of SB623 cells at 7
days
post-TBI, (2) animals subjected to TBI followed by infusion of vehicle at 7
days post-
TBI, and (3) control sham-operated age-matched adult Sprague-Dawley rats (n=3
per
group). Zymographic assays of the laser-captured biobridges from animals
subjected
to TBI revealed two-fold and nine-fold upregulation of matrix
metalloproteinase 9
(MMP-9) expression/activity- in animals that received SB623 cell transplants,
compared to vehicle-infused animals or sham-operated animals, at one month and
three months post-transplantation, respectively (Example 11).
[0043] MMPs have been implicated in recovery in chronic brain
injury29, and
inhibition of MMP activity has been shown to abrogate migration of neurogenic
cells
from the SVZ into damaged tissues and to retard neurovascular remodeling30.
MMPs
may thus play a role in facilitating host cell migration towards injured brain
areas as
part of the process by which SB623 cells provide functional recovery from TBI.
[00441 In summary, the inventors have discovered that transplantation
of
SB623 cells remodeled the traumatically injured brain by creating a biobridge
between the SVZ and the pen-injured cortex. This method of cell therapy can
now be
used to create similar biobridges between neurogenic and non-neurogenic sites,
to
facilitate injury-specific migration of cells across tissues that might
otherwise pose
barriers to cell motility.
Formulations, kits and routes of administration
[0045] Therapeutic compositions comprising SB623 cells as disclosed
herein
are also provided. Such compositions typically comprise the SB623 cells and a
pharmaceutically acceptable carrier. Supplementary active compounds can also
be
incorporated into SB623 cell compositions.
13

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
[0046] The therapeutic compositions disclosed herein are useful for,
inter alia,
treating TBI and modulating stem cell migration in the brain. Accordingly, a
"therapeutically effective amount" of a composition comprising SB623 cells is
any
amount that reduces symptoms of TBI or that stimulates miuration of stem cells
in the
brain. For example, dosage amounts can vary from about 100; 500; 1,000; 2,500;
5,000; 10, 000; 20,000; 50,000; 100,000; 300,000; 500,000; 1,000,000;
5,000,000 to
10,000,000 cells or more (or any integral value therebetween); with a
frequency of
administration of, e.g., once per day, twice per week, once per week, twice
per month,
once per month, depending upon, e.g., body weight, route of administration,
severity
of disease, etc. Thus, a therapeutically effective amount can comprise a
plurality of
administrations of the same amount, or different amounts, of SB623 cells. In
certain
embodiments, a single administration of SB623 cells is a therapeutically
effective
amount.
[0047] Various pharmaceutical compositions and techniques for their
preparation and use are known to those of skill in the art in light of the
present
disclosure. For a detailed listing of suitable pharmacological compositions
and
techniques for their administration one may refer to texts such as Remington's
Pharmaceutical Sciences, 17th ed. 1985; Brunton et al., "Goodman and Gilman's
The
Phatinacological Basis of Therapeutics," McGraw-Hill, 2005; University of the
Sciences in Philadelphia (eds.), "Remington: The Science and Practice of
Pharmacy,"
Lippincott Williams & Wilkins, 2005; and University of the Sciences in
Philadelphia
(eds.), "Remington: The Principles of Pharmacy Practice," Lippincott Williams
&
Wilkins, 2008.
[0048] The cells described herein may be suspended in a
physiologically
compatible carrier for transplantation. As used herein, the telin
"physiologically
compatible carrier" refers to a carrier that is compatible with the SB623
cells and with
any other ingredients of the formulation, and is not deleterious to the
recipient thereof.
Those of skill in the art are familiar with physiologically compatible
carriers.
Examples of suitable carriers include cell culture medium (e.g Eagle's minimal
essential medium), phosphate buffered saline, Hank's balanced salt solution+/-
glucose
(HBSS), and multiple electrolyte solutions such as, e.g., Plasma-LyteTM A
(Baxter).
[0049] The volume of a SB623 cell suspension administered to a subject
will
vary depending on the site of transplantation, treatment goal and number of
cells in
solution. Typically the amount of cells administered will be a therapeutically
14

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
effective amount. As used herein, a "therapeutically effective amount" or
"effective
amount" refers to the number of transplanted cells which are required to
effect
treatment of the particular disorder; i.e., to produce a reduction in the
amount and/or
severity of the symptoms associated with that disorder. For example, in the
case of
TBI, transplantation of a therapeutically effective amount of SB623 cells
results in
reduction and/or reversal of the symptoms of TBI; e.g., restoration of
locomotor
activity and neurological perfaimance, and stimulation of migration of host
neurogenic cells. Therapeutically effective amounts vary with the type and
extent of
brain damage, and can also vary depending on the overall condition of the
subject.
[0050] The disclosed therapeutic compositions can also include
pharmaceutically acceptable materials, compositions or vehicle, such as a
liquid or
solid filler, diluent, excipicnt, solvent or encapsulating material, i.e.,
carriers. These
carriers can, for example, stabilize the SB623 cells and/or facilitate the
survival of the
SB623 cells in the body. Each carrier should be "acceptable" in the sense of
being
compatible with the other ingredients of the formulation and not injurious to
the
subject. Some examples of materials which can serve as pharmaceutically-
acceptable
carriers include: sugars, such as lactose, glucose and sucrose; starches, such
as corn
starch and potato starch; cellulose and its derivatives, such as sodium
carboxymethyl
cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt;
gelatin;
talc; excipients, such as cocoa butter and suppository waxes; oils, such as
peanut oil,
cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean
oil; glycols,
such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and
polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar;
buffering
agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid;
pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol;
phosphate buffer
solutions; and other non-toxic compatible substances employed in
pharmaceutical
formulations. Wetting agents, emulsifiers and lubricants, such as sodium
lauryl
sulfate and magnesium stearate, as well as coloring agents, release agents,
coating
agents, sweetening, flavoring and perfuming agents, preservatives and
antioxidants
can also be present in the compositions.
100511 Exemplary formulations include, but are not limited to, those
suitable
for parenteral administration, e.g., intrapulmonary, intravenous, intra-
arterial, intra-
ocular, intra-cranial, sub-meningial, or subcutaneous administration,
including
formulations encapsulated in micelles, liposomes or drug-release capsules
(active

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
agents incorporated within a biocompatible coating designed for slow-release);
ingestible formulations; formulations for topical use, such as eye drops,
creams,
ointments and gels; and other formulations such as inhalants, aerosols and
sprays.
The dosage of the compositions of the disclosure will vary according to the
extent and
severity of the need for treatment, the activity of the administered
composition, the
general health of the subject, and other considerations well known to the
skilled
artisan.
[0052] In additional embodiments, the compositions described herein
are
delivered intracranially at or near a site of traumatic brain injury. Such
localized
delivery allows for the delivery of the composition non-systemically, thereby
reducing the body burden of the composition as compared to systemic delivery.
Local
delivery can be achieved, for example, by intra-cranial injection, or through
the use of
various medically implanted devices including, but not limited to, stents and
catheters,
or can be achieved by inhalation, phlebotomy, or surgery. Methods for coating,
implanting, embedding, and otherwise attaching desired agents to medical
devices
such as stents and catheters are established in the art and contemplated
herein.
[0053] Another aspect of the present disclosure relates to kits for
carrying out
the administration of SB623 cells, optionally in combination with another
therapeutic
agent, to a subject. In one embodiment, a kit comprises a composition of SB623
cells,
formulated in a pharmaceutical carrier, suitable for transplantation.
EXAMPLES
[0054] In the studies disclosed herein, rats were subjected to
experimental
traumatic brain injury (TBI) and, seven days later, those having sufficient
locomotor
and neurological deficits received transplants of either SB623 cells or
vehicle to the
injured area. Values of locomotor and neurological performance were evaluated
prior
to TBI (baseline values), again at 7 days after TBI (prior to
transplantation), and
monthly thereafter for 3 months after TBI.
[0055] Following completion of behavioral testing at 1 month and 3
months
after TBI, randomly selected animals were euthanized (n=10 per group) by
transeardial perfusion with 4% paraformaldehyde. Their brains were removed and
sectioned for evaluation of persistence of the transplanted cells,
histological
appearance of brain tissue in and around the injured area, expression of
various neural
16

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
markers in and around the injured area, and zymogenic activity in and around
the
injured area,
[0056] Transplant outcomes were evaluated using the following
criteria: 1)
locomotor behavior via elevated body swing test (EBST) and Rotorod; (2)
neurological performance via a Bederson-modified neurological examination; 3)
lesion volume via histology (H&E stained sections); 4) graft survival via
immunohistochemistry using an antibody (HuNu) that specifically detects human
cells, and; 5) mechanism-based immunohistochemial analyses of neuroprotection
and/or regeneration using antibodies directed against the grafted human cells
and host
cells.
Example 1: Preparation of MSCs and SB623 cells
[0057] Bone marrow aspirates from adult human donors were obtained
from
Lonza Walkersville, Inc. (Walkersville, MD) and plated in a-MEM (Mediatech,
Herndon, VA) supplemented with 10% fetal bovine serum (Hyclone, Logan, UT), 2
mM L-glutamine (Invitrogen, Carlsbad, CA) and penicillin/streptomycin
(Invitrogen).
Cells were cultured for three days at 37 C and 5% CO2, to obtain a monolayer
of
adherent cells. After removal of non-adherent cells, culture was continued
under the
same conditions for two weeks. During this time, cells were passaged twice,
using
0.25% trypsin/EDTA. A portion of the cells from the second passage were frozen
as
MSCs.
[0058] The remaining cells from the second passage were plated and
transfected, using Fugene6 (Roche Diagnostics, Indianapolis, IN), with a
plasmid
containing sequences encoding a Notch intracellular domain operatively linked
to a
cytomegalovirus promoter (pCMV-ENICD1-SV40-NeoR). This plasmid also
contained sequences encoding resistance to neomycin and G418 under the
transcriptional control of a SV40 promoter. Transfected cells were cultured at
37 C
and 5% CO2 in the growth medium described in the previous paragraph,
supplemented with 100 i.g/m1 G418 (Invitrogen, Carlsbad, CA). After seven
days,
G418-resistant colonies were expanded and the culture was passaged twice.
After the
second passage, the cells were collected and frozen as SB623 cells.
[0059] MSCs and SB623 cells prepared as described herein were thawed
as
required and used for further study.
17

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
Example 2: Induction of TBI in a rat model
[0060] A total of 40 animals identified at baseline (prior to TBI
surgery) as
exhibiting normal behaviors (50-60% bias swing activity in EBST; 60 seconds
staying
time on Rotorod; and a mean Bederson score of at most 0-0.5), received TBI
surgery
as described below.
[0061] All surgical procedures were conducted under aseptic
conditions.
Adult male Sprague-Dawley rats were anesthetized with 1.5% isofluorane and
checked for pain reflexes. Under deep anesthesia, animals underwent a moderate
TBI
model, as follows. Each animal was placed in a stereotaxic frame, with
anesthesia
being maintained with 1-2% isofluorane administered via a gas mask. After
exposing
the skull, a 4-mm craniectomy was performed over the left frontoparietal
cortex, with
its center at ¨2.0 mm AP and +2.0 mm ML to the bregma. A pneumatically
operated
metal impactor, with a diameter of 3 mm, was used to impact the brain at a
velocity of
6.0 m/s, reaching a depth of 1.0 mm below the dura mater layer and remaining
in the
brain for 150 milliseconds. The impactor rod was angled 15 to the vertical,
so as to
be perpendicular to the tangential plane of the brain surface at the impact
site. A
linear variable displacement transducer (Macro sensors, Pennsauken, NJ)
connected to
the impactor was used to measure velocity and duration, to verify consistency.
[0062] Subsequent to controlled cortical impact injury, the incision
was
sutured after bleeding ceased. An integrated heating pad and rectal
thermometer unit
with feedback control allowed maintenance of body temperature at normal
limits. All
animals were monitored until recovery from anesthesia. In addition, animals
were
weighed and observed daily for three consecutive days following induction of
TBI.
weighed twice a week thereafter, and monitored daily throughout the study for
health
status and any signs that indicated problems or complications.
Example 3: Grafting of SB623 Cells
[0063] Of the animals subjected to TBI, only those having the
following
degree of behavioral impairment at Day 7 post-TBI were selected for
transplantation
studies: at least 75% bias swing activity in the EBST; 30 seconds or less
staying time
on the Rotorod; and a mean Bederson score of at least 2.5. Those animals that
were
selected were randomly assigned either to a group receiving SB623 transplants
(n=20)
or to a group receiving vehicle infusion (n=20). The target area for
transplantation
18

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
was the medial cortex, which corresponded to the pen-injured cortical area
based on
previously established target sites for similar stereotaxic implants.
[0064] All surgical procedures were conducted under aseptic
conditions.
Animals were anesthetized with 1.5% isofluorane and checked for pain reflexes.
Once deep anesthesia was achieved (as determined by the loss of pain reflex),
the hair
was shaved around the area of the surgical incision (skull area), leaving
enough
border to prevent contamination of the operative site. This was followed by
two
surgical germicidal scrubs of the site, and draping with sterile drapes.
[0065] The animal was then fixed to a stereotaxic apparatus (Kopf
Instruments, Tujunga, CA), and a small opening was made in the skull with a
bun.
The coordinates of the opening were 0.5 mm anterior and 1.0 mm lateral to the
bregma and 2.0 mm below the dural surface; these were selected to correspond
to the
cortical area adjacent to the core injury site, based on the atlas of Paxinos
and Watson
(1998). A 26-gauge Hamilton syringe, containing test material, was then
lowered into
the opening. With a single needle pass, 3 deposits of 3 ul each were made.
Each
deposit consisted of 100,000 viable cells in 3 ul of Plasmalyte A, infused
over a
period of 3 minutes. Following an additional 2-minute absorption time, the
needle
was retracted and the wound was closed with a stainless steel wound clip. A
heating
pad and a rectal thermometer allowed maintenance of body temperature at about
37 C
throughout surgery and following recovery from anesthesia. Control injections
contained Plasmalyte A only.
[0066] Treated and control animals were subjected to elevated body
swing test
(EBST, Example 4), neurological examination (Example 5), and the Rotorod test
(Example 6) at baseline (prior to TBI), at 7 days after TBI (just prior to
transplantation) and monthly thereafter up to 3 months post-TBI.
[0067] In addition, brains of treated and control animals were
characterized
histologically at one and three months post-TBI to determine degree of damage
(Examples 8 and 9); the extent of proliferation, migration and neural
differentiation of
host cells (Example 10); and the presence of zymogenic activity (Example 11).
Example 4: Elevated Body Swing Test (EBST)
[0068] All investigators testing the animals were blinded to the
treatment
condition. The EBST was conducted by handling the animal by its tail and
recording
the direction in which the animal swung its head. The test apparatus consisted
of a
19

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
clear Plexiglas box (40 x 40 x 35.5 cm). The animal was gently picked up at
the base
of the tail, and elevated by the tail until the animal's nose was at a height
of 2 inches
(5 cm) above the surface. The direction of the swing (left or right) was
recorded once
the animal's head moved sideways approximately 10 degrees from the midline
position of the body. After a single swing, the animal was placed back in the
Plexiglas box and allowed to move freely for 30 seconds prior to retesting.
These
steps were repeated for a total of 20 assays for each animal. Uninjured rats
display a
50% swing bias, that is, the same number of swings to the left and to the
right. A
75% swing bias indicated 15 swings in one direction and 5 in the other during
20
trials. Previous results utilizing the EBST have indicated that unilaterally
lesioned
animals display >75% biased swing activity at one month after a nigrostriatal
lesion
or unilateral hemispheric injury; and that such asymmetry is stable for up to
six
months3'26.
[0069] The results of the EBST are shown in Figure 1. After TBI,
essentially
all animals exhibited biased swing activity. In animals transplanted with
SB623 cells,
biased swing activity steadily decreased over the three-month period following
TBI
and transplantation. By contrast, in animals transplanted with vehicle, the
percentage
of animals exhibiting biased swing activity after TBI remained essentially
unchanged.
Example 5: Modified Bederson Neurological Examination
[0070] About one hour after conclusion of the EBST, a modified
Bederson-
Neurological exam was conducted, following the procedures previously
describee26
with minor modifications. Neurologic score for each rat was obtained using 3
tests
which included (1) forelimb retraction, which measured the ability of the
animal to
........................................................... replace the
forelimb after it was displaced laterally by 2 to 3 cm, graded from 0
(immediate replacement) to 3 (replacement after several seconds or no
replacement);
(2) beam walking ability, graded 0 for a rat that readily traversed a 2.4-cm-
wide, 80-
cm-long beam to 3 for a rat unable to stay on the beam for 10 seconds; and (3)
bilateral forepaw grasp, which measured the ability to hold onto a 2-mm-
diameter
steel rod, graded 0 for a rat with noimal forepaw grasping behavior to 3 for a
rat
unable to grasp with the forepaws. The scores from all 3 tests, which were
conducted
over a period of about 15 minutes on each assessment day, were added to give a
mean
neurologic deficit score (maximum possible score. 9 points divided by 3 tests
= 3).

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
[00711 The results of these neurological examinations are shown in
Figure 2.
After TBI, the mean neurological score was 2.5 (out of 3) in all animals. In
animals
transplanted with SB623 cells, this score was steadily reduced (indicating
improved
neurological function) over the three-month period following TBI and
transplantation.
Improvement of neurological function in animals transplanted with SB623 cells
was
statistically significant (p < 0.05) compared to animals that had been infused
with
vehicle.
Example 6: Rotorod Test
[0072] One hour after completion of the neurological exam, the animals were
subjected to the Rotorod test. This test involved placement of the animal on
a
rotating treadmill that accelerates from 4 rpm to 40 rpm over a 60-second
period
(Rotorod , Accuscan, Inc., Columbus, OH). The total number of seconds an
animal
was able to remain on the treadmill was recorded and used as an index of motor
coordination. Previous results using a TBI model system have shown that
injured
animals were able to remain on the Rotorod for significantly shorter times,
compared
to sham-operated or normal controls.
[0073] The results of this assay are shown in Figure 3. Uninjured
animals
were able to remain on the treadmill for an average of 60 seconds. The mean
time on
the treadmill fell to below 20 seconds seven days after TBI. In animals
transplanted
with SB623 cells after TBI, mean time on the treadmill doubled to
approximately 40
seconds. These improvements were statistically significant compared to animals
subjected to TBI that had been infused with vehicle.
Example 7: Perfusion and sectioning
[0074] At 1 month and 3 months after TBI, following completion of
behavioral testing as described in Examples 4-6 , randomly-selected rats were
euthanized (n=10 per group) by transcardial perfusion with 4%
parafoimaldehyde.
The brains were dissected, post-fixed overnight in 4% paraformaldehyde, then
immersed in 30% sucrose. Beginning at bregma-5.2 mm anteriorly, each forebrain
was cut into 40 um coronal sections, moving posteriorly until bregma-8.8mm.
Sections were processed for determinations of brain damage and analysis of
cell
survival in the pen-lesion area as described in Examples 8 and 9.
21

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
Example 8: Measurement of Brain Damage
[0075] Preparation and examination of brain sections was undertaken to
identify the extent of brain damage and host cell survival. At least 4 coronal
tissue
sections per brain were processed for hematoxylin and eosin (H&E) or Nissl
staining.
Cerebral damage was quantitated by determining the indirect lesion area, which
was
calculated by subtracting the intact area of the ipsilateral hemisphere from
the area of
the contralateral hemisphere. The lesion volume was presented as a volume
percentage of the lesion compared to the contralateral hemisphere, by summing
lesion
areas from serial sections.
[0076] The results, shown quantitatively in Figure 4B, indicate that
animals
subjected to TBI that received transplants of SB623 cells experienced
significantly
less damage to the cortical core and the pen-injured cortical areas, compared
to
animals subjected to TBI that received infusion of vehicle.
Example 9: Analysis of Cell Survival in the pen-TB! Lesion Area
[0077] Randomly selected high power fields, corresponding to the pen-
injured
cortical area, were examined to count surviving host cells in this region.
Results are
shown in Figure 4A.
Example 10: Immunohistoehemistry
[0078] Floating sections were processed for immunofluorescent
microscopy.
Briefly, 40 I..tm cryostat sectioned tissues were examined at 4X magnification
and
digitized using a PC-based Image Tools computer program. Engraftment of
transplanted SB623 cells was assessed using monoclonal human specific antibody
HuNu that did not cross-react with rodent proteins. Additional brain sections
were
processed for mechanism-based immunohistochemical analyses of brain tissue
samples focusing on cell proliferation (Ki67), migration (doublecortin or DCX)
and
neural differentiation (nestin). Brain sections were blind-coded and
Abercrombie's
formula was used to calculate the total number of immunopositive cells3'26.
[0079] The results of these analyses showed that transplantation of SB623
cells induced formation of a biobridge between the SVZ and the impacted cortex
consisting of highly proliferative, neurally committed and migratory cells. At
one
month post-TBI, immunofluorescent and confocal microscopy revealed a surge of
endogenous cell proliferation (evidenced by cells expressing Ki67) and
immature
22

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
neural differentiation (cells expressing nestin) in the pen-injured cortical
areas and
subventricular zone (SVZ), with a stream of migrating cells (cells expressing
doublecortin) along the corpus callosum (CC) of the animals that had received
transplants of SB623 cells. Brains from animals that had received vehicle
alone
displayed limited cell proliferation and neural differentiation, and scattered
migration
in the pen-injured cortical areas, with almost no newly formed cells present
in the
SVZ. At three months post- FBI, the brains from SB623-transplanted animals
exhibited much more massive cell proliferation and neural differentiation
encasing the
pen-injured cortical areas, accompanied by a solid stream of neuronally
labeled cells
(expressing both nestin and doublecortin) migrating, not just along, but
across the CC
from the SVZ to the impacted cortex. By contrast, in brains from vehicle-
infused
animals, cell proliferation was enhanced, but the newly formed cells were
"trapped"
within the SVZ and the CC and only a few cells were able to reach the impacted
cortex. Quantitative analyses of Ki67, nestin and DCX immunolabeled cells in
SVZ,
CC and CTX revealed statistically significant differences between transplanted
and
vehicle-infused animals (Figures 5-7).
Example 11: Zymography
100801 A separate cohort of animals from that whose analysis was
described
in Examples 4-10 was used to test for the presence and/or activity of
proteolytic
enzymes after transplantation of SB623 cells into injured brain. Rats were
subjected
to TBI, then transplanted with either SB623 cells or vehicle. A control group
of age-
matched sham-operated adult Sprague-Dawley rats was subjected to the same
experimental procedure (n=3 rats per group). At one month and three months
after
TBI, tissue corresponding to the biobridge fottned by the cells migrating from
the
SVZ to the impacted cortex was obtained by laser dissection. After extraction,
the
tissue was placed in cryotubes and flash frozen in liquid nitrogen. The tubes
were
stored in a -80 C freezer until homogenization.
[0081] Samples were homogenized in 450 [IL of cold working buffer
containing 50 mM Tris-HC1 (pH 7.5), 75 mM NaC1, and 1 mM PMSF. The tissue
was processed with a homogenizer for 10 minutes and centrifuged at 4 C for 20
minutes at 13000 rpm. The supernatants were separated, frozen and kept at -80
C
until use. The total protein concentration in the supernatant was assessed by
the
Bradford method.
23

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
[0082] On the day that zymography was conducted, a volume equivalent
to 50
ug of total protein was loaded into a freshly prepared gelatin zymography gel.
All
gels contained a control lane that was loaded with 0.5 ng recombinant MMP-9,
which
was used as a standard for both enzyme amount (in ng) and gelatinolytic
activity
(expressed as relative optical density units, see below). Proteins were
electrophoretieally separated in the gel under non-reducing conditions at 100
V. After
electrophoresis the gels were washed in 125 ml 2.5% Triton twice for 20
minutes.
The gels were then incubated in activation buffer (Zymogram Development
Buffer,
Bio-Rad, Hercules, CA) for 20 hours at 37 C. The next day, the gels were
stained
with Coomassie Blue R-250 Staining Solution (Bio-Rad) for 3 hours and
destained for
25 minutes with Destain Solution (Bio-Rad). The gelatinolytic activity of the
samples
was assessed by densitometric analysis (Gel-Pro v 3.1, Media Cybernetics,
Carlsbad,
CA) of the bands. The molecular weights of proteins in regions of the gel
exhibiting
lytic activity were determined by comparison to pre-stained standard protein
marker
(Bio-Rad) run on the same gel. Activity was expressed as optical density
relative to
that of 0.5 ng of recombinant MMP-9, which was run in the gel as a standard.
[0083] The results are shown in Figure 8. The laser-captured
biobridges
(corresponding to brain tissue between the SVZ and the impacted cortex) from
animals transplanted with SB623 cells after TBI expressed high levels of MMP-9
gelatinolytic activity at one month and three months post-TBI. The levels in
SB623-
treated animals were significantly higher than those in biobridges from
vehicle-
infused and sham-operated animals (p <0.05) at both time points. Although
biobridges from vehicle-infused animals showed a significant increase in MMP-9
activity compared to sham-operated animals at one month post-TBI, these levels
reverted to control levels (i.e., not significantly different from those of
sham-operated
animals) at three months post-TBI.
[0084] For detection on blots, membranes were blocked with blotting
grade
non-fat dry milk (Bio-Rad). After washing with 0.1% tween 20- tri s- buffered
saline
(TTBS), the membranes were incubated with 1 ug/ml anti MMP-9 monoclonal mouse
antibody overnight at 4 C. Membranes were washed again in TTBS, incubated with
secondary antibody (1:1,000 dilution of horseradish peroxidase-conjugated goat
anti-
mouse IgG, Calbiochem) for one hour and finally developed with horseradish
peroxidase development solution (ECL advance detection kit, ,kmersham). The
membranes were exposed to autoradiography films (Hyblot CL, Denville
Scientific
24

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
Inc.). The density of the sample bands for the zymograms was expressed as
maximal
optical density relative to the standard band (0.5 ng recombinant MMP-9).

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
REFERENCES
1. Joyner, A. L. et al. Production of a mutation in mouse En-2 gene by
homologous recombination in embryonic stem cells. Nature 338, 153-156 (1989)
2. Yasuhara, T. et at. Transplantation of human neural stem cells exerts
neuroprotection in a rat model of Parkinson's disease. I Neurosci. 26, 12497-
12511
(2006)
3. Yasuhara, T. et at. Intravenous grafts recapitulate the neurorestoration
afforded by intracerebrally delivered multipotent adult progenitor cells in
neonatal
hypoxic-ischemic rats. I Cereb. Blood Flow Metab. 28, 1804-1810 (2008)
4. Borlongan, C. V. et at. Central nervous system entry of peripherally
injected umbilical cord blood cells is not required for neuroprotection in
stroke.
Stroke 35, 2385-2389 (2004)
5. Pastori, C. et at. Arterially perfused neurosphere-derived cells distribute
outside the ischemic core in a model of transient focal ischemia and
reperfusion in
vitro. PLoS One 3, e2754 (2008)
6. Redmond, D. E. Jr. et al. Behavioral improvement in a primate Parkinson's
model is associated with multiple homeostatic effects of human neural stem
cells.
Proc. Natl. Acad Sci. U. S. A. 104, 12175-12180 (2007)
7. Lee, J. P. et al. Stem cells act through multiple mechanisms to benefit
mice
with neurodegenerative metabolic disease. Nat. Med. 13, 439-447 (2007)
8. Ma, D. K. et al. Epigenetic choreographers of neurogenesis in the adult
mammalian brain. Nat. Neurosci. 13, 1338-1344 (2010)
9. Hong, S. H. et at. Cell fate potential of human pluripotent stem cells is
encoded by histone modifications. Cell Stern Cell. 9, 24-36 (2011)
10. Kim, Y. et at. Mouse B-type lamins are required for proper organogenesis
but not by embryonic stem cells. Science. 334, 1706-1710 (2011)
11. Borlongan, C. V. Bone marrow stem cell mobilization in stroke: a
'bonehead' may be good after all! Leukemia 25, 1674-1686 (2011)
12. Barha, C. K. et al. Progesterone treatment normalizes the levels of cell
proliferation and cell death in the dentate gyrus of the hippocampus after
traumatic
brain injury. Exp. Neurol. 231, 72-81 (2011)
13. Jaskelioff, M. et at. Telomerase reactivation reverses tissue degeneration
in aged telomerase-deficient mice. Nature 469, 102-106 (2011)
26

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
14. Wang, L. et al. Tumor necrosis factor a primes cerebral endothelial cells
for erythropoietin-induced angiogenesis. 1 Cereb. Blood Flow Metab. 31, 640-
647
(2011)
15. Andres, R. H. et at. Human neural stem cells enhance structural plasticity
and axonal transport in the ischaemic brain. Brain. 134, 1777-1789 (2011)
16. Liu, Z. et at. Bone marrow stromal cells promote skilled motor recovery
and enhance contralesional axonal connections after ischemic stroke in adult
mice.
Stroke 42, 740-744 (2011)
17. Mazzocchi-Jones, D. et al. Embryonic striatal grafts restore bi-
directional
synaptic plasticity in a rodent model of Huntington's disease. Eur. 1
Neurosci. 30,
2134-2142 (2009)
18. Lee, H. S. et al. Foxa2 and Nurrl synergistically yield A9 nigral
dopamine neurons exhibiting improved differentiation, function, and cell
survival.
Stem Cells 28, 501-512 (2010)
19. Harps, G. et al. Differentiated Parkinson patient-derived induced
pluripotent stern cells grow in the adult rodent brain and reduce motor
asymmetry in
Parkinsonian rats. Proc. Natl. Acad. Sci. U S. A. 107, 15921-15926 (2010)
20. Yasuda, A. et at. Significance of remyelination by neural stem/progenitor
cells transplanted into the injured spinal cord. Stem Cells. 29, 1983-1994
(2011)
21. Mezey, E. The therapeutic potential of bone marrow-derived stem cells.
Biochem. 112, 2683-2687 (2011)
22. Sanai, N. et al. Corridors of migrating neurons in the human brain and
their decline during infancy. Nature 478, 382-386 (2011)
23. Carlen, 1\4. et al. Forebrain ependymal cells are Notch-dependent and
generate neuroblasts and astrocytes after stroke. Nat. Neurosci. 12, 259-267
(2009)
24. Robel, S. et al. The stem cell potential of glia: lessons from reactive
gliosis. Nat. Rev. Neurosci. 12, 88-104 (2011)
25. Seol, H. J. et at. Genetically engineered human neural stem cells with
rabbit carboxyl esterase can target brain metastasis from breast cancer.
Cancer Lett.
311, 152-159 (2011)
26. Yasuhara, T. et al. Notch-induced rat and human bone marrow stromal
cell grafts reduce ischemic cell loss and ameliorate behavioral deficits in
chronic
stroke animals. Stem Cells Dev. 18, 1501-1514 (2009)
27

CA 02872188 2019-10-30
WO 2013/172944
PCT/US2013/030895
27. Pollock, K. et at. A conditionally immortal clonal stem cell line form
human cortical neuroepithelium for the treatment of ischemic stroke. Exp.
Neural.
199, 143-155 (2006)
28. Dezawa, M. et al. Specific induction of neuronal cells from bone marrow
stromal cells and application for autologous transplantation. J Clin Invest.
113, 1701-
(2004)
29. Zhao, B.Q., Tejima, E., Lo, E.11. Neurovascular proteases in brain injury,
hemorrhage and remodeling after stroke. Stroke. 38, 748-752 (2007)
30. Zhao, B. Q. et al. Role of matrix metalloproteinases in delayed cortical
10 responses after stroke. Nat Med. 12,441-445 (2006)
28

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Grant by Issuance 2018-08-14
Inactive: Cover page published 2018-08-13
Inactive: Final fee received 2018-06-27
Pre-grant 2018-06-27
Notice of Allowance is Issued 2018-01-16
Letter Sent 2018-01-16
4 2018-01-16
Notice of Allowance is Issued 2018-01-16
Change of Address or Method of Correspondence Request Received 2018-01-09
Inactive: Q2 passed 2018-01-07
Inactive: Approved for allowance (AFA) 2018-01-07
Amendment Received - Voluntary Amendment 2017-09-14
Inactive: S.30(2) Rules - Examiner requisition 2017-08-14
Inactive: Report - No QC 2017-08-11
Amendment Received - Voluntary Amendment 2017-04-25
Inactive: S.30(2) Rules - Examiner requisition 2016-12-30
Inactive: Report - QC passed 2016-12-29
Amendment Received - Voluntary Amendment 2016-05-09
Inactive: S.30(2) Rules - Examiner requisition 2015-11-12
Inactive: Report - No QC 2015-11-04
Inactive: IPC deactivated 2015-01-24
Inactive: Cover page published 2015-01-15
Inactive: IPC assigned 2015-01-14
Inactive: IPC removed 2015-01-14
Inactive: IPC assigned 2015-01-14
Inactive: IPC assigned 2015-01-14
Inactive: First IPC assigned 2015-01-14
Correct Applicant Requirements Determined Compliant 2015-01-09
Inactive: Acknowledgment of national entry - RFE 2015-01-09
Letter Sent 2014-12-02
Inactive: First IPC assigned 2014-11-28
Inactive: Notice - National entry - No RFE 2014-11-28
Inactive: IPC assigned 2014-11-28
Inactive: IPC assigned 2014-11-28
Inactive: IPC assigned 2014-11-28
Inactive: IPC assigned 2014-11-28
Application Received - PCT 2014-11-28
All Requirements for Examination Determined Compliant 2014-11-14
Request for Examination Requirements Determined Compliant 2014-11-14
Request for Examination Received 2014-11-14
Correct Applicant Request Received 2014-11-06
Inactive: Correspondence - PCT 2014-11-06
National Entry Requirements Determined Compliant 2014-10-30
Application Published (Open to Public Inspection) 2013-11-21

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2018-02-21

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
UNIVERSITY OF SOUTH FLORIDA
SANBIO, INC.
Past Owners on Record
CASEY C. CASE
CESAR V. BORLONGAN
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2014-10-29 28 1,668
Abstract 2014-10-29 2 110
Drawings 2014-10-29 7 933
Claims 2014-10-29 2 51
Representative drawing 2015-01-14 1 68
Cover Page 2015-01-14 1 100
Description 2016-05-08 30 1,683
Claims 2016-05-08 4 114
Description 2017-04-24 30 1,576
Claims 2017-04-24 4 113
Claims 2017-09-13 3 99
Cover Page 2018-07-17 1 30
Maintenance fee payment 2024-02-19 49 2,016
Acknowledgement of Request for Examination 2014-12-01 1 176
Reminder of maintenance fee due 2014-11-30 1 111
Notice of National Entry 2014-11-27 1 193
Notice of National Entry 2015-01-08 1 203
Commissioner's Notice - Application Found Allowable 2018-01-15 1 162
PCT 2014-10-29 5 167
Correspondence 2014-11-05 4 144
Examiner Requisition 2015-11-11 5 278
Amendment / response to report 2016-05-08 16 670
Examiner Requisition 2016-12-29 3 218
Amendment / response to report 2017-04-24 13 585
Examiner Requisition 2017-08-13 4 188
Amendment / response to report 2017-09-13 5 162
Final fee 2018-06-26 1 42